Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 819
Filtrar
1.
Neuron ; 112(16): 2721-2731.e5, 2024 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-38901431

RESUMEN

The ventral tegmental area (VTA) is a critical node in circuits governing motivated behavior and is home to diverse populations of neurons that release dopamine, gamma-aminobutyric acid (GABA), glutamate, or combinations of these neurotransmitters. The VTA receives inputs from many brain regions, but a comprehensive understanding of input-specific activation of VTA neuronal subpopulations is lacking. To address this, we combined optogenetic stimulation of select VTA inputs with single-nucleus RNA sequencing (snRNA-seq) and highly multiplexed in situ hybridization to identify distinct neuronal clusters and characterize their spatial distribution and activation patterns. Quantification of immediate-early gene (IEG) expression revealed that different inputs activated select VTA subpopulations, which demonstrated cell-type-specific transcriptional programs. Within dopaminergic subpopulations, IEG induction levels correlated with differential expression of ion channel genes. This new transcriptomics-guided circuit analysis reveals the diversity of VTA activation driven by distinct inputs and provides a resource for future analysis of VTA cell types.


Asunto(s)
Genes Inmediatos-Precoces , Optogenética , Área Tegmental Ventral , Área Tegmental Ventral/metabolismo , Área Tegmental Ventral/citología , Animales , Ratones , Optogenética/métodos , Neuronas Dopaminérgicas/metabolismo , Masculino , Neuronas/metabolismo , Ratones Endogámicos C57BL , Análisis de Secuencia de ARN/métodos
2.
Nature ; 630(8017): 677-685, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38839962

RESUMEN

All drugs of abuse induce long-lasting changes in synaptic transmission and neural circuit function that underlie substance-use disorders1,2. Another recently appreciated mechanism of neural circuit plasticity is mediated through activity-regulated changes in myelin that can tune circuit function and influence cognitive behaviour3-7. Here we explore the role of myelin plasticity in dopaminergic circuitry and reward learning. We demonstrate that dopaminergic neuronal activity-regulated myelin plasticity is a key modulator of dopaminergic circuit function and opioid reward. Oligodendroglial lineage cells respond to dopaminergic neuronal activity evoked by optogenetic stimulation of dopaminergic neurons, optogenetic inhibition of GABAergic neurons, or administration of morphine. These oligodendroglial changes are evident selectively within the ventral tegmental area but not along the axonal projections in the medial forebrain bundle nor within the target nucleus accumbens. Genetic blockade of oligodendrogenesis dampens dopamine release dynamics in nucleus accumbens and impairs behavioural conditioning to morphine. Taken together, these findings underscore a critical role for oligodendrogenesis in reward learning and identify dopaminergic neuronal activity-regulated myelin plasticity as an important circuit modification that is required for opioid reward.


Asunto(s)
Analgésicos Opioides , Vaina de Mielina , Vías Nerviosas , Plasticidad Neuronal , Recompensa , Área Tegmental Ventral , Animales , Femenino , Masculino , Ratones , Analgésicos Opioides/farmacología , Dopamina/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/efectos de los fármacos , Ratones Endogámicos C57BL , Morfina/farmacología , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/metabolismo , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/fisiología , Núcleo Accumbens/citología , Núcleo Accumbens/metabolismo , Núcleo Accumbens/fisiología , Núcleo Accumbens/efectos de los fármacos , Oligodendroglía/metabolismo , Oligodendroglía/citología , Oligodendroglía/efectos de los fármacos , Optogenética , Área Tegmental Ventral/fisiología , Área Tegmental Ventral/citología , Área Tegmental Ventral/efectos de los fármacos , Vías Nerviosas/efectos de los fármacos , Linaje de la Célula
3.
Nat Commun ; 15(1): 4233, 2024 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-38762463

RESUMEN

The ventral pallidum (VP) contains GABA and glutamate neurons projecting to ventral tegmental area (VTA) whose stimulation drives approach and avoidance, respectively. Yet little is known about the mechanisms by which VP cell types shape VTA activity and drive behavior. Here, we found that both VP GABA and glutamate neurons were activated during approach to reward or by delivery of an aversive stimulus. Stimulation of VP GABA neurons inhibited VTA GABA, but activated dopamine and glutamate neurons. Remarkably, stimulation-evoked activation was behavior-contingent such that VTA recruitment was inhibited when evoked by the subject's own action. Conversely, VP glutamate neurons activated VTA GABA, as well as dopamine and glutamate neurons, despite driving aversion. However, VP glutamate neurons evoked dopamine in aversion-associated ventromedial nucleus accumbens (NAc), but reduced dopamine release in reward-associated dorsomedial NAc. These findings show how heterogeneous VP projections to VTA can be engaged to shape approach and avoidance behaviors.


Asunto(s)
Reacción de Prevención , Prosencéfalo Basal , Neuronas GABAérgicas , Ácido Glutámico , Recompensa , Área Tegmental Ventral , Área Tegmental Ventral/fisiología , Área Tegmental Ventral/metabolismo , Área Tegmental Ventral/citología , Animales , Ácido Glutámico/metabolismo , Prosencéfalo Basal/metabolismo , Prosencéfalo Basal/fisiología , Masculino , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/fisiología , Reacción de Prevención/fisiología , Ratones , Dopamina/metabolismo , Núcleo Accumbens/metabolismo , Núcleo Accumbens/citología , Núcleo Accumbens/fisiología , Neuronas/metabolismo , Neuronas/fisiología , Ácido gamma-Aminobutírico/metabolismo , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/fisiología , Ratones Endogámicos C57BL , Conducta Animal/fisiología
4.
Nature ; 630(8015): 141-148, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38778097

RESUMEN

Fentanyl is a powerful painkiller that elicits euphoria and positive reinforcement1. Fentanyl also leads to dependence, defined by the aversive withdrawal syndrome, which fuels negative reinforcement2,3 (that is, individuals retake the drug to avoid withdrawal). Positive and negative reinforcement maintain opioid consumption, which leads to addiction in one-fourth of users, the largest fraction for all addictive drugs4. Among the opioid receptors, µ-opioid receptors have a key role5, yet the induction loci of circuit adaptations that eventually lead to addiction remain unknown. Here we injected mice with fentanyl to acutely inhibit γ-aminobutyric acid-expressing neurons in the ventral tegmental area (VTA), causing disinhibition of dopamine neurons, which eventually increased dopamine in the nucleus accumbens. Knockdown of µ-opioid receptors in VTA abolished dopamine transients and positive reinforcement, but withdrawal remained unchanged. We identified neurons expressing µ-opioid receptors in the central amygdala (CeA) whose activity was enhanced during withdrawal. Knockdown of µ-opioid receptors in CeA eliminated aversive symptoms, suggesting that they mediate negative reinforcement. Thus, optogenetic stimulation caused place aversion, and mice readily learned to press a lever to pause optogenetic stimulation of CeA neurons that express µ-opioid receptors. Our study parses the neuronal populations that trigger positive and negative reinforcement in VTA and CeA, respectively. We lay out the circuit organization to develop interventions for reducing fentanyl addiction and facilitating rehabilitation.


Asunto(s)
Fentanilo , Receptores Opioides mu , Refuerzo en Psicología , Animales , Femenino , Masculino , Ratones , Analgésicos Opioides/farmacología , Analgésicos Opioides/administración & dosificación , Núcleo Amigdalino Central/citología , Núcleo Amigdalino Central/efectos de los fármacos , Núcleo Amigdalino Central/metabolismo , Dopamina/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Fentanilo/farmacología , Ratones Endogámicos C57BL , Núcleo Accumbens/citología , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Trastornos Relacionados con Opioides/metabolismo , Trastornos Relacionados con Opioides/patología , Optogenética , Receptores Opioides mu/metabolismo , Síndrome de Abstinencia a Sustancias/metabolismo , Síndrome de Abstinencia a Sustancias/patología , Área Tegmental Ventral/citología , Área Tegmental Ventral/efectos de los fármacos , Área Tegmental Ventral/metabolismo
5.
J Neurosci ; 44(18)2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38485256

RESUMEN

The ventral pallidum (VP) is a central hub in the reward circuitry with diverse projections that have different behavioral roles attributed mostly to the connectivity with the downstream target. However, different VP projections may represent, as in the striatum, separate neuronal populations that differ in more than just connectivity. In this study, we performed in mice of both sexes a multimodal dissection of four major projections of the VP-to the lateral hypothalamus (VP→LH), ventral tegmental area (VP→VTA), lateral habenula (VP→LHb), and mediodorsal thalamus (VP→MDT)-with physiological, anatomical, genetic, and behavioral tools. We also tested for physiological differences between VP neurons receiving input from nucleus accumbens medium spiny neurons (MSNs) that express either the D1 (D1-MSNs) or the D2 (D2-MSNs) dopamine receptor. We show that each VP projection (1) when inhibited during a cocaine conditioned place preference (CPP) test affects performance differently, (2) receives a different pattern of inputs using rabies retrograde labeling, (3) shows differentially expressed genes using RNA sequencing, and (4) has projection-specific characteristics in excitability and synaptic input characteristics using whole-cell patch clamp. VP→LH and VP→VTA projections have different effects on CPP and show low overlap in circuit tracing experiments, as VP→VTA neurons receive more striatal input, while VP→LH neurons receive more olfactory input. Additionally, VP→VTA neurons are less excitable, while VP→LH neurons are more excitable than the average VP neuron, a difference driven mainly by D2-MSN-responding neurons. Thus, VP→VTA and VP→LH neurons may represent largely distinct populations of VP neurons.


Asunto(s)
Prosencéfalo Basal , Cocaína , Vías Nerviosas , Recompensa , Animales , Ratones , Prosencéfalo Basal/fisiología , Masculino , Cocaína/farmacología , Cocaína/administración & dosificación , Femenino , Vías Nerviosas/fisiología , Ratones Endogámicos C57BL , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D1/genética , Receptores de Dopamina D2/metabolismo , Receptores de Dopamina D2/genética , Área Tegmental Ventral/fisiología , Área Tegmental Ventral/citología
7.
Science ; 383(6678): 55-61, 2024 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-38175903

RESUMEN

Decision-making is always coupled with some level of risk, with more pathological forms of risk-taking decisions manifesting as gambling disorders. In macaque monkeys trained in a high risk-high return (HH) versus low risk-low return (LL) choice task, we found that the reversible pharmacological inactivation of ventral Brodmann area 6 (area 6V) impaired the risk dependency of decision-making. Selective optogenetic activation of the mesofrontal pathway from the ventral tegmental area (VTA) to the ventral aspect of 6V resulted in stronger preference for HH, whereas activation of the pathway from the VTA to the dorsal aspect of 6V led to LL preference. Finally, computational decoding captured the modulations of behavioral preference. Our results suggest that VTA inputs to area 6V determine the decision balance between HH and LL.


Asunto(s)
Asunción de Riesgos , Área Tegmental Ventral , Animales , Área Tegmental Ventral/citología , Área Tegmental Ventral/fisiología , Macaca fuscata
8.
Nature ; 619(7969): 332-337, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37380765

RESUMEN

Fast-acting neurotransmitters and slow, modulatory neuropeptides are co-released from neurons in the central nervous system, albeit from distinct synaptic vesicles1. The mechanisms of how co-released neurotransmitters and neuropeptides that have opposing actions-for example, stimulatory versus inhibitory-work together to exert control of neural circuit output remain unclear. This has been difficult to resolve owing to the inability to selectively isolate these signalling pathways in a cell- and circuit-specific manner. Here we developed a genetic-based anatomical disconnect procedure that utilizes distinct DNA recombinases to independently facilitate CRISPR-Cas9 mutagenesis2 of neurotransmitter- and neuropeptide-related genes in distinct cell types in two different brain regions simultaneously. We demonstrate that neurons within the lateral hypothalamus that produce the stimulatory neuropeptide neurotensin and the inhibitory neurotransmitter GABA (γ-aminobutyric acid) utilize these signals to coordinately activate dopamine-producing neurons of the ventral tegmental area. We show that GABA release from lateral hypothalamus neurotensin neurons inhibits GABA neurons within the ventral tegmental area, disinhibiting dopamine neurons and causing a rapid rise in calcium, whereas neurotensin directly generates a slow inactivating calcium signal in dopamine neurons that is dependent on the expression of neurotensin receptor 1 (Ntsr1). We further show that these two signals work together to regulate dopamine neuron responses to maximize behavioural responding. Thus, a neurotransmitter and a neuropeptide with opposing signals can act on distinct timescales through different cell types to enhance circuit output and optimize behaviour.


Asunto(s)
Encéfalo , Vías Nerviosas , Neurotensina , Neurotransmisores , Transducción de Señal , Encéfalo/citología , Encéfalo/metabolismo , Calcio/metabolismo , Sistemas CRISPR-Cas , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Neuronas GABAérgicas , Ácido gamma-Aminobutírico/metabolismo , Edición Génica , Área Hipotalámica Lateral/citología , Área Hipotalámica Lateral/metabolismo , Neurotensina/metabolismo , Neurotransmisores/metabolismo , Receptores de Neurotensina/metabolismo , Área Tegmental Ventral/citología , Área Tegmental Ventral/metabolismo
9.
Nature ; 608(7922): 374-380, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35831501

RESUMEN

Food and water are rewarding in part because they satisfy our internal needs1,2. Dopaminergic neurons in the ventral tegmental area (VTA) are activated by gustatory rewards3-5, but how animals learn to associate these oral cues with the delayed physiological effects of ingestion is unknown. Here we show that individual dopaminergic neurons in the VTA respond to detection of nutrients or water at specific stages of ingestion. A major subset of dopaminergic neurons tracks changes in systemic hydration that occur tens of minutes after thirsty mice drink water, whereas different dopaminergic neurons respond to nutrients in the gastrointestinal tract. We show that information about fluid balance is transmitted to the VTA by a hypothalamic pathway and then re-routed to downstream circuits that track the oral, gastrointestinal and post-absorptive stages of ingestion. To investigate the function of these signals, we used a paradigm in which a fluid's oral and post-absorptive effects can be independently manipulated and temporally separated. We show that mice rapidly learn to prefer one fluid over another based solely on its rehydrating ability and that this post-ingestive learning is prevented if dopaminergic neurons in the VTA are selectively silenced after consumption. These findings reveal that the midbrain dopamine system contains subsystems that track different modalities and stages of ingestion, on timescales from seconds to tens of minutes, and that this information is used to drive learning about the consequences of ingestion.


Asunto(s)
Dopamina , Neuronas Dopaminérgicas , Hipotálamo , Vías Nerviosas , Nutrientes , Estado de Hidratación del Organismo , Área Tegmental Ventral , Animales , Señales (Psicología) , Digestión , Dopamina/metabolismo , Neuronas Dopaminérgicas/fisiología , Ingestión de Alimentos , Tracto Gastrointestinal/metabolismo , Hipotálamo/citología , Hipotálamo/fisiología , Mesencéfalo/citología , Mesencéfalo/fisiología , Ratones , Nutrientes/metabolismo , Estado de Hidratación del Organismo/efectos de los fármacos , Recompensa , Factores de Tiempo , Área Tegmental Ventral/citología , Área Tegmental Ventral/fisiología , Agua/metabolismo , Agua/farmacología , Equilibrio Hidroelectrolítico
10.
Nature ; 608(7922): 368-373, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35896744

RESUMEN

Ketamine is used clinically as an anaesthetic and a fast-acting antidepressant, and recreationally for its dissociative properties, raising concerns of addiction as a possible side effect. Addictive drugs such as cocaine increase the levels of dopamine in the nucleus accumbens. This facilitates synaptic plasticity in the mesolimbic system, which causes behavioural adaptations and eventually drives the transition to compulsion1-4. The addiction liability of ketamine is a matter of much debate, in part because of its complex pharmacology that among several targets includes N-methyl-D-aspartic acid (NMDA) receptor (NMDAR) antagonism5,6. Here we show that ketamine does not induce the synaptic plasticity that is typically observed with addictive drugs in mice, despite eliciting robust dopamine transients in the nucleus accumbens. Ketamine nevertheless supported reinforcement through the disinhibition of dopamine neurons in the ventral tegmental area (VTA). This effect was mediated by NMDAR antagonism in GABA (γ-aminobutyric acid) neurons of the VTA, but was quickly terminated by type-2 dopamine receptors on dopamine neurons. The rapid off-kinetics of the dopamine transients along with the NMDAR antagonism precluded the induction of synaptic plasticity in the VTA and the nucleus accumbens, and did not elicit locomotor sensitization or uncontrolled self-administration. In summary, the dual action of ketamine leads to a unique constellation of dopamine-driven positive reinforcement, but low addiction liability.


Asunto(s)
Ketamina , Trastornos Relacionados con Sustancias , Animales , Dopamina/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Ketamina/efectos adversos , Ketamina/farmacología , Ratones , Plasticidad Neuronal/efectos de los fármacos , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/metabolismo , Refuerzo en Psicología , Autoadministración , Trastornos Relacionados con Sustancias/etiología , Trastornos Relacionados con Sustancias/prevención & control , Área Tegmental Ventral/citología , Área Tegmental Ventral/efectos de los fármacos
11.
Stem Cell Reports ; 16(11): 2718-2735, 2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34678205

RESUMEN

In Parkinson's disease (PD), substantia nigra (SN) dopaminergic (DA) neurons degenerate, while related ventral tegmental area (VTA) DA neurons remain relatively unaffected. Here, we present a methodology that directs the differentiation of mouse and human pluripotent stem cells toward either SN- or VTA-like DA lineage and models their distinct vulnerabilities. We show that the level of WNT activity is critical for the induction of the SN- and VTA-lineage transcription factors Sox6 and Otx2, respectively. Both WNT signaling modulation and forced expression of these transcription factors can drive DA neurons toward the SN- or VTA-like fate. Importantly, the SN-like lineage enriched DA cultures recapitulate the selective sensitivity to mitochondrial toxins as observed in PD, while VTA-like neuron-enriched cultures are more resistant. Furthermore, a proteomics approach led to the identification of compounds that alter SN neuronal survival, demonstrating the utility of our strategy for disease modeling and drug discovery.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Degeneración Nerviosa/genética , Enfermedad de Parkinson/genética , Células Madre Pluripotentes/metabolismo , Sustancia Negra/metabolismo , Área Tegmental Ventral/metabolismo , Animales , Diferenciación Celular/genética , Línea Celular , Neuronas Dopaminérgicas/citología , Células Madre Embrionarias Humanas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Modelos Neurológicos , Células Madre Embrionarias de Ratones/metabolismo , Factores de Transcripción Otx/genética , Factores de Transcripción Otx/metabolismo , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Células Madre Pluripotentes/citología , Factores de Transcripción SOXD/genética , Factores de Transcripción SOXD/metabolismo , Sustancia Negra/citología , Área Tegmental Ventral/citología
12.
Mol Psychiatry ; 26(11): 6170-6186, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34642456

RESUMEN

Plasticity of neurons in the ventral tegmental area (VTA) is critical for establishment of drug dependence. However, the remodeling of the circuits mediating the transition between positive and negative effect remains unclear. Here, we used neuronal activity-dependent labeling technique to characterize and temporarily control the VTA neuronal ensembles recruited by the initial morphine exposure (morphine-positive ensembles, Mor-Ens). Mor-Ens preferentially projected to NAc, and induced dopamine-dependent positive reinforcement. Electrophysiology and rabies viral tracing revealed the preferential connections between the VTA-projective corticotrophin-releasing hormone (CRH) neurons of central amygdala (CRHCeA→VTA) and Mor-Ens, which was enhanced after escalating morphine exposure and mediated the negative effect during opiate withdrawal. Pharmacologic intervention or CRISPR-mediated repression of CRHR1 in Mor-Ens weakened the inhibitory CRHCeA→VTA inputs, and alleviated the negative effect during opiate withdrawal. These data suggest that neurons encoding opioid reward experience are inhibited by enhanced CRHCeA→VTA inputs induced by chronic morphine exposure, leading to negative effect during opiate withdrawal, and provide new insight into the pathological changes in VTA plasticity after drug abuse and mechanism of opiate dependence.


Asunto(s)
Morfina/efectos adversos , Plasticidad Neuronal , Síndrome de Abstinencia a Sustancias/fisiopatología , Área Tegmental Ventral , Amígdala del Cerebelo/citología , Analgésicos Opioides/efectos adversos , Animales , Hormona Liberadora de Corticotropina/metabolismo , Área Tegmental Ventral/citología , Área Tegmental Ventral/fisiopatología
13.
Psychoneuroendocrinology ; 133: 105393, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34481327

RESUMEN

Maternal separation (MS) is a known chronic stressor in the postnatal period and when associated with another paradigm like the activity-based anorexia (ABA) rat model, causes different effects in the two sexes. In ABA females, the separation leads to increased hyperactivity and anxiety reduction, whereas, in males, the separation induces decreased locomotor activity without similar reduction of anxiety-like behaviors as observed in females. To understand the mechanisms altered by MS in synergy with the induction of the anorexic-like phenotype, we considered the reward system, which involves neurons synthesizing dopamine (DA) in the ventral tegmental area (VTA), substantia nigra pars compacta, and serotoninergic neurons in the dorsal raphe nucleus. Moreover, we analyzed the orexin circuit in the lateral hypothalamic area (LHA), which affects DA synthesis in the VTA and is also known to regulate food consumption and locomotor activity. Rats of both sexes were exposed to the two paradigms (MS and ABA), leading to four experimental groups for each sex: non-separated control (CON), non-separated ABA groups (ABA), MS control (MSCON), and MS plus ABA groups (MSABA). Immunohistochemistry analysis was performed to determine quantitative differences in the number of cells expressing DA, orexin, and serotonin (5-HT) among the experimental groups. The results showed that, in the DA system, the effect of MS was more evident in females than in males, with a substantial increase in DA cells in the VTA of MSABA. However, the analysis of the orexin system revealed a similar cellular increment in the LHA in the non-separated ABA groups of both sexes. Regarding 5-HT, there was an opposite effect in males and females of the MSABA groups, with only females showing a greater density of 5-HT cells. The changes in the reward system could partially explain the behavioral data: the hyperactivity, weight loss, and decreased anxiety levels of the MSABA females could be linked to an increase in DA and 5-HT cells, whereas in males, MS could mitigate the behavioral effects of the ABA protocol affecting the anxiety levels and locomotor activity through a lack of increased activation of the reward system.


Asunto(s)
Anorexia , Privación Materna , Recompensa , Animales , Anorexia/complicaciones , Ansiedad/complicaciones , Modelos Animales de Enfermedad , Dopamina , Núcleo Dorsal del Rafe/citología , Femenino , Masculino , Neuronas , Orexinas , Porción Compacta de la Sustancia Negra/citología , Ratas , Serotonina , Área Tegmental Ventral/citología
14.
Neuropharmacology ; 197: 108746, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34371079

RESUMEN

Increasing evidence indicates that the melanocortin and mesolimbic dopamine (DA) systems interact to regulate feeding and body weight. Because melanocortin-3 receptors (MC3R) are highly expressed in the ventral tegmental area (VTA), we tested whether VTA neurons expressing these receptors (VTA MC3R neurons) control feeding and body weight in vivo. We also tested whether there were sex differences in the ability of VTA MC3R neurons to control feeding, as MC3R -/- mice show sex-dependent alterations in reward feeding and DA levels, and there are clear sex differences in multiple DA-dependent behaviors and disorders. Designer receptors exclusively activated by designer drugs (DREADD) were used to acutely activate and inhibit VTA MC3R neurons and changes in food intake and body weight were measured. Acutely altering the activity of VTA MC3R neurons decreased feeding in an activity- and sex-dependent manner, with acute activation decreasing feeding, but only in females, and acute inhibition decreasing feeding, but only in males. These differences did not appear to be due to sex differences in the number of VTA MC3R neurons, the ability of hM3Dq to activate VTA MC3R neurons, or the proportion of VTA MC3R neurons expressing tyrosine hydroxylase (TH). These studies demonstrate an important role for VTA MC3R neurons in the control of feeding and reveal important sex differences in behavior, whereby opposing changes in neuronal activity in male and female mice cause similar changes in behavior.


Asunto(s)
Actividad Motora/fisiología , Neuronas/fisiología , Receptor de Melanocortina Tipo 3/fisiología , Área Tegmental Ventral/fisiología , Animales , Peso Corporal , Drogas de Diseño/farmacología , Dopamina/metabolismo , Conducta Alimentaria , Femenino , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Receptor de Melanocortina Tipo 3/genética , Recompensa , Caracteres Sexuales , Área Tegmental Ventral/citología
16.
Sci Rep ; 11(1): 12873, 2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-34145364

RESUMEN

The reward system, which consists of dopaminergic neurons projecting from the ventral tegmental area (VTA) to the nucleus accumbens and caudate-putamen in the striatum, has an important role in the pathogenesis of not only drug addiction but also diet-induced obesity. In the present study, we examined whether signaling through glucocorticoid receptors (GRs) in the reward system affects the rewarding value of a high-fat diet (HFD). To do so, we generated mice that lack functional GRs specifically in dopaminergic neurons (D-KO mice) or corticostriatal neurons (CS-KO mice), subjected the mice to caloric restriction stress conditions, and evaluated the rewarding value of a HFD by conditioned place preference (CPP) test. Caloric restriction induced increases in serum corticosterone to similar levels in all genotypes. While CS-KO as well as WT mice exhibited a significant preference for HFD in the CPP test, D-KO mice exhibited no such preference. There were no differences between WT and D-KO mice in consumption of HFD after fasting or cognitive function evaluated by a novel object recognition test. These data suggest that glucocorticoid signaling in the VTA increases the rewarding value of a HFD under restricted caloric stress.


Asunto(s)
Dieta Alta en Grasa , Neuronas/metabolismo , Receptores de Glucocorticoides/metabolismo , Recompensa , Transducción de Señal , Área Tegmental Ventral/metabolismo , Animales , Condicionamiento Clásico , Neuronas Dopaminérgicas/metabolismo , Metabolismo Energético , Ayuno , Expresión Génica , Ratones , Ratones Transgénicos , Modelos Animales , Receptores de Glucocorticoides/genética , Área Tegmental Ventral/citología
17.
Nat Commun ; 12(1): 2811, 2021 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-33990558

RESUMEN

The supramammillary region (SuM) is a posterior hypothalamic structure, known to regulate hippocampal theta oscillations and arousal. However, recent studies reported that the stimulation of SuM neurons with neuroactive chemicals, including substances of abuse, is reinforcing. We conducted experiments to elucidate how SuM neurons mediate such effects. Using optogenetics, we found that the excitation of SuM glutamatergic (GLU) neurons was reinforcing in mice; this effect was relayed by their projections to septal GLU neurons. SuM neurons were active during exploration and approach behavior and diminished activity during sucrose consumption. Consistently, inhibition of SuM neurons disrupted approach responses, but not sucrose consumption. Such functions are similar to those of mesolimbic dopamine neurons. Indeed, the stimulation of SuM-to-septum GLU neurons and septum-to-ventral tegmental area (VTA) GLU neurons activated mesolimbic dopamine neurons. We propose that the supramammillo-septo-VTA pathway regulates arousal that reinforces and energizes behavioral interaction with the environment.


Asunto(s)
Neuronas Dopaminérgicas/fisiología , Hipotálamo Posterior/citología , Hipotálamo Posterior/fisiología , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Conducta Consumatoria/efectos de los fármacos , Conducta Consumatoria/fisiología , Dopamina/fisiología , Femenino , Ácido Glutámico/fisiología , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Neurológicos , Vías Nerviosas/citología , Vías Nerviosas/fisiología , Optogenética , Ratas , Ratas Wistar , Refuerzo en Psicología , Tabique del Cerebro/citología , Tabique del Cerebro/efectos de los fármacos , Tabique del Cerebro/fisiología , Área Tegmental Ventral/citología , Área Tegmental Ventral/fisiología , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/administración & dosificación
18.
Sci Rep ; 11(1): 8706, 2021 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-33888815

RESUMEN

The ventral tegmental area (VTA) is the origin of dopaminergic neurons and the dopamine (DA) reward pathway. This pathway has been widely studied in addiction and drug reinforcement studies and is believed to be the central processing component of the reward circuit. In this study, we used a well-established rat model to expose mother dams to alcohol, nicotine-alcohol, and saline perinatally. DA and non-DA neurons collected from the VTA of the rat pups were used to study expression profiles of miRNAs and mRNAs. miRNA pathway interactions, putative miRNA-mRNA target pairs, and downstream modulated biological pathways were analyzed. In the DA neurons, 4607 genes were differentially upregulated and 4682 were differentially downregulated following nicotine-alcohol exposure. However, in the non-DA neurons, only 543 genes were differentially upregulated and 506 were differentially downregulated. Cell proliferation, differentiation, and survival pathways were enriched after the treatments. Specifically, in the PI3K/AKT signaling pathway, there were 41 miRNAs and 136 mRNAs differentially expressed in the DA neurons while only 16 miRNAs and 20 mRNAs were differentially expressed in the non-DA neurons after the nicotine-alcohol exposure. These results depicted that chronic nicotine and alcohol exposures during pregnancy differentially affect both miRNA and gene expression profiles more in DA than the non-DA neurons in the VTA. Understanding how the expression signatures representing specific neuronal subpopulations become enriched in the VTA after addictive substance administration helps us to identify how neuronal functions may be altered in the brain.


Asunto(s)
Neuronas Dopaminérgicas/efectos de los fármacos , Etanol/administración & dosificación , Exposición Materna , Nicotina/administración & dosificación , Área Tegmental Ventral/efectos de los fármacos , Animales , Neuronas Dopaminérgicas/metabolismo , Femenino , Masculino , MicroARNs/metabolismo , Embarazo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Área Tegmental Ventral/citología , Área Tegmental Ventral/metabolismo
19.
Learn Mem ; 28(4): 104-108, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33723029

RESUMEN

The dopamine system has been implicated in decision-making particularly when associated with effortful behavior. We examined acute optogenetic stimulation of dopamine cells in the ventral tegmental area (VTA) as mice engaged in an effort-based decision-making task. Tyrosine hydroxylase-Cre mice were injected with Cre-dependent ChR2 or eYFP control virus in the VTA. While eYFP control mice showed effortful discounting, stimulation of dopamine cells in ChR2 mice disrupted effort-based decision-making by reducing choice toward the lever associated with a preferred outcome and greater effort. Surprisingly, disruptions in effortful discounting were observed in subsequent test sessions conducted in the absence of optogenetic stimulation, however during these sessions ChR2 mice displayed enhanced high choice responding across trial blocks. These findings suggest increases in VTA dopamine cell activity can disrupt effort-based decision-making in distinct ways dependent on the timing of optogenetic stimulation.


Asunto(s)
Conducta Animal/fisiología , Toma de Decisiones/fisiología , Neuronas Dopaminérgicas/fisiología , Área Tegmental Ventral/fisiología , Animales , Condicionamiento Operante/fisiología , Ratones , Optogenética , Desempeño Psicomotor/fisiología , Tirosina 3-Monooxigenasa , Área Tegmental Ventral/citología
20.
PLoS One ; 16(2): e0245663, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33534843

RESUMEN

Parkinson's disease is associated with the loss of dopamine (DA) neurons in ventral mesencephalon. We have previously reported that no single neurotrophic factor we tested protected DA neurons from the dopaminergic toxin 1-methyl-4-phenylpyridinium (MPP+) in dissociated cultures isolated from the P0 rat substantia nigra, but that a combination of five neurotrophic factors was protective. We now report that cerebral DA neurotrophic factor (CDNF) and a variant of neurturin (NRTN), N4, were also not protective when provided alone but were protective when added together. In cultures isolated from the substantia nigra, MPP+ (10 µM) decreased tyrosine hydroxylase-positive cells to 41.7 ± 5.4% of vehicle control. Although treatment of cultures with 100 ng/ml of either CDNF or N4 individually before and after toxin exposure did not significantly increase survival in MPP+-treated cultures, when the two trophic factors were added together at 100 ng/ml each, survival of cells was increased 28.2 ± 6.1% above the effect of MPP+ alone. In cultures isolated from the ventral tegmental area, another DA rich area, a higher dose of MPP+ (1 mM) was required to produce an EC50 in TH-positive cells but, as in the substantia nigra, only the combination of CDNF and N4 (100 ng/ml each) was successful at increasing the survival of these cells compared to MPP+ alone (by 22.5 ± 3.5%). These data support previous findings that CDNF and N4 may be of therapeutic value for treatment of PD, but suggest that they may need to be administered together.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Mesencéfalo/metabolismo , Factores de Crecimiento Nervioso/farmacología , Fármacos Neuroprotectores/farmacología , Neurturina/farmacología , 1-Metil-4-fenilpiridinio , Animales , Células CHO , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cricetulus , Dopamina/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Humanos , Nomifensina/farmacología , Ratas Sprague-Dawley , Sustancia Negra/citología , Tritio/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Área Tegmental Ventral/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA